1
|
Li H, Seugnet L. Decoding the nexus: branched-chain amino acids and their connection with sleep, circadian rhythms, and cardiometabolic health. Neural Regen Res 2025; 20:1350-1363. [PMID: 39075896 PMCID: PMC11624887 DOI: 10.4103/nrr.nrr-d-23-02020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/18/2024] [Accepted: 05/12/2024] [Indexed: 07/31/2024] Open
Abstract
The sleep-wake cycle stands as an integrative process essential for sustaining optimal brain function and, either directly or indirectly, overall body health, encompassing metabolic and cardiovascular well-being. Given the heightened metabolic activity of the brain, there exists a considerable demand for nutrients in comparison to other organs. Among these, the branched-chain amino acids, comprising leucine, isoleucine, and valine, display distinctive significance, from their contribution to protein structure to their involvement in overall metabolism, especially in cerebral processes. Among the first amino acids that are released into circulation post-food intake, branched-chain amino acids assume a pivotal role in the regulation of protein synthesis, modulating insulin secretion and the amino acid sensing pathway of target of rapamycin. Branched-chain amino acids are key players in influencing the brain's uptake of monoamine precursors, competing for a shared transporter. Beyond their involvement in protein synthesis, these amino acids contribute to the metabolic cycles of γ-aminobutyric acid and glutamate, as well as energy metabolism. Notably, they impact GABAergic neurons and the excitation/inhibition balance. The rhythmicity of branched-chain amino acids in plasma concentrations, observed over a 24-hour cycle and conserved in rodent models, is under circadian clock control. The mechanisms underlying those rhythms and the physiological consequences of their disruption are not fully understood. Disturbed sleep, obesity, diabetes, and cardiovascular diseases can elevate branched-chain amino acid concentrations or modify their oscillatory dynamics. The mechanisms driving these effects are currently the focal point of ongoing research efforts, since normalizing branched-chain amino acid levels has the ability to alleviate the severity of these pathologies. In this context, the Drosophila model, though underutilized, holds promise in shedding new light on these mechanisms. Initial findings indicate its potential to introduce novel concepts, particularly in elucidating the intricate connections between the circadian clock, sleep/wake, and metabolism. Consequently, the use and transport of branched-chain amino acids emerge as critical components and orchestrators in the web of interactions across multiple organs throughout the sleep/wake cycle. They could represent one of the so far elusive mechanisms connecting sleep patterns to metabolic and cardiovascular health, paving the way for potential therapeutic interventions.
Collapse
Affiliation(s)
- Hui Li
- Department of Neurology, Xijing Hospital, Xi’an, Shaanxi Province, China
| | - Laurent Seugnet
- Centre de Recherche en Neurosciences de Lyon, Integrated Physiology of the Brain Arousal Systems (WAKING), Université Claude Bernard Lyon 1, INSERM U1028, CNRS UMR 5292, Bron, France
| |
Collapse
|
2
|
Hong W, You G, Luo Z, Zhang M, Chen J. High gestational leucine level dampens WDPCP/MAPK signaling to impair the EMT and migration of cardiac microvascular endothelial cells in congenital heart defects. Pulm Circ 2024; 14:e70013. [PMID: 39582775 PMCID: PMC11582015 DOI: 10.1002/pul2.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 10/08/2024] [Accepted: 11/01/2024] [Indexed: 11/26/2024] Open
Abstract
Congenital heart defects (CHDs) represent one of the most prevalent categories of neonatal defects, and maternal dietary patterns have been linked to the risk of these conditions. Branched-chain amino acids (BCAAs), particularly leucine, are essential for various metabolic and physiological processes involved in heart development. In this study, we examined the molecular mechanisms through which elevated levels of leucine induce defects in cardiac microvascular endothelial cells. We collected plasma samples from healthy controls and neonatal patients with CHDs, employed a high-leucine diet for pregnant female mice, and applied high-leucine treatment in human cardiac microvascular endothelial cells (HCMECs). The impacts of high-leucine levels on WD Repeat Containing Planar Cell Polarity Effector (WDPCP)/MAPK signaling axis were investigated in the cell and animal models. We reported heightened plasma leucine levels in neonatal patients with CHDs and observed that a high-leucine diet in pregnant mice was associated with reduced expression of WDPCP and attenuated MAPK/ERK signaling. High-leucine treatment in HCMECs impaired epithelial-mesenchymal transition (EMT) and cell migration; however, overexpression of WDPCP or activation of MAPK exhibited a rescue effect. The upregulation of endomucin (EMCN) under high-leucine conditions contributed to the impaired EMT and migratory capacity of HCMECs, and the WDPCP/MAPK signaling axis regulated EMCN overexpression in response to high-leucine treatment. High levels of leucine in neonatal patients with CHDs may inhibit the WDPCP/MAPK axis, leading to an increase in EMCN expression that undermines the function of cardiac microvascular endothelial cells. These findings suggest the potential of targeting the WDPCP/MAPK axis as an intervention strategy for neonatal CHDs.
Collapse
Affiliation(s)
- Wei Hong
- Kunming Children's HospitalKunmingYunnanChina
| | - Guozhou You
- Kunming Children's HospitalKunmingYunnanChina
| | | | | | - Jian Chen
- Kunming Children's HospitalKunmingYunnanChina
| |
Collapse
|
3
|
Nguyen DC, Wells CK, Taylor MS, Martinez-Ondaro Y, Brittian KR, Brainard RE, Moore IV JB, Hill BG. Dietary Branched Chain Amino Acids Modify Post-Infarct Cardiac Remodeling and Function in the Murine Heart. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.12.603348. [PMID: 39071416 PMCID: PMC11275808 DOI: 10.1101/2024.07.12.603348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Introduction Branch-chain amino acids (BCAA) are markedly elevated in the heart following myocardial infarction (MI) in both humans and animal models. Nevertheless, it remains unclear whether dietary BCAA levels influence post-MI remodeling. We hypothesize that lowering dietary BCAA levels prevents adverse cardiac remodeling after MI. Methods and Results To assess whether altering dietary BCAA levels would impact circulating BCAA concentrations, mice were fed a low (1/3×), normal (1×), or high (2×) BCAA diet over a 7-day period. We found that mice fed the low BCAA diet had >2-fold lower circulating BCAA concentrations when compared with normal and high BCAA diet feeding strategies; notably, the high BCAA diet did not further increase BCAA levels over the normal chow diet. To investigate the impact of dietary BCAAs on cardiac remodeling and function after MI, male and female mice were fed either the low or high BCAA diet for 2 wk prior to MI and for 4 wk after MI. Although body weights or heart masses were not different in female mice fed the custom diets, male mice fed the high BCAA diet had significantly higher body and heart masses than those on the low BCAA diet. Echocardiographic assessments revealed that the low BCAA diet preserved stroke volume and cardiac output for the duration of the study, while the high BCAA diet led to progressive decreases in cardiac function. Although no discernible differences in cardiac fibrosis, scar collagen topography, or cardiomyocyte cross-sectional area were found between the dietary groups, male mice fed the high BCAA diet showed longer cardiomyocytes and higher capillary density compared with the low BCAA group. Conclusions Provision of a diet low in BCAAs to mice mitigates eccentric cardiomyocyte remodeling and loss of cardiac function after MI, with dietary effects more prominent in males.
Collapse
Affiliation(s)
- Daniel C. Nguyen
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY
- Department of Physiology, University of Louisville, Louisville, KY
| | - Collin K. Wells
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY
| | - Madison S. Taylor
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY
| | - Yania Martinez-Ondaro
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY
| | - Kenneth R. Brittian
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY
| | | | - Joseph B. Moore IV
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY
| | - Bradford G. Hill
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY
| |
Collapse
|
4
|
Festus ID, Spilberg J, Young ME, Cain S, Khoshnevis S, Smolensky MH, Zaheer F, Descalzi G, Martino TA. Pioneering new frontiers in circadian medicine chronotherapies for cardiovascular health. Trends Endocrinol Metab 2024; 35:607-623. [PMID: 38458859 DOI: 10.1016/j.tem.2024.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 03/10/2024]
Abstract
Cardiovascular disease (CVD) is a global health concern. Circadian medicine improves cardiovascular care by aligning treatments with our body's daily rhythms and their underlying cellular circadian mechanisms. Time-based therapies, or chronotherapies, show special promise in clinical cardiology. They optimize treatment schedules for better outcomes with fewer side effects by recognizing the profound influence of rhythmic body cycles. In this review, we focus on three chronotherapy areas (medication, light, and meal timing) with potential to enhance cardiovascular care. We also highlight pioneering research in the new field of rest, the gut microbiome, novel chronotherapies for hypertension, pain management, and small molecules that targeting the circadian mechanism.
Collapse
Affiliation(s)
- Ifene David Festus
- Centre for Cardiovascular Investigations, University of Guelph; Guelph, Ontario, Canada; Department of Biomedical Sciences, University of Guelph; Guelph, Ontario, Canada
| | - Jeri Spilberg
- Centre for Cardiovascular Investigations, University of Guelph; Guelph, Ontario, Canada; Department of Biomedical Sciences, University of Guelph; Guelph, Ontario, Canada
| | - Martin E Young
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sean Cain
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, Victoria, Australia
| | - Sepideh Khoshnevis
- Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Michael H Smolensky
- Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, USA; Department of Internal Medicine, Division of Cardiology, McGovern School of Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Fariya Zaheer
- Department of Biomedical Sciences, University of Guelph; Guelph, Ontario, Canada
| | - Giannina Descalzi
- Department of Biomedical Sciences, University of Guelph; Guelph, Ontario, Canada
| | - Tami A Martino
- Centre for Cardiovascular Investigations, University of Guelph; Guelph, Ontario, Canada; Department of Biomedical Sciences, University of Guelph; Guelph, Ontario, Canada.
| |
Collapse
|
5
|
Tanase DM, Valasciuc E, Costea CF, Scripcariu DV, Ouatu A, Hurjui LL, Tarniceriu CC, Floria DE, Ciocoiu M, Baroi LG, Floria M. Duality of Branched-Chain Amino Acids in Chronic Cardiovascular Disease: Potential Biomarkers versus Active Pathophysiological Promoters. Nutrients 2024; 16:1972. [PMID: 38931325 PMCID: PMC11206939 DOI: 10.3390/nu16121972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Branched-chain amino acids (BCAAs), comprising leucine (Leu), isoleucine (Ile), and valine (Val), are essential nutrients vital for protein synthesis and metabolic regulation via specialized signaling networks. Their association with cardiovascular diseases (CVDs) has become a focal point of scientific debate, with emerging evidence suggesting both beneficial and detrimental roles. This review aims to dissect the multifaceted relationship between BCAAs and cardiovascular health, exploring the molecular mechanisms and clinical implications. Elevated BCAA levels have also been linked to insulin resistance (IR), type 2 diabetes mellitus (T2DM), inflammation, and dyslipidemia, which are well-established risk factors for CVD. Central to these processes are key pathways such as mammalian target of rapamycin (mTOR) signaling, nuclear factor kappa-light-chain-enhancer of activate B cells (NF-κB)-mediated inflammation, and oxidative stress. Additionally, the interplay between BCAA metabolism and gut microbiota, particularly the production of metabolites like trimethylamine-N-oxide (TMAO), adds another layer of complexity. Contrarily, some studies propose that BCAAs may have cardioprotective effects under certain conditions, contributing to muscle maintenance and metabolic health. This review critically evaluates the evidence, addressing the biological basis and signal transduction mechanism, and also discusses the potential for BCAAs to act as biomarkers versus active mediators of cardiovascular pathology. By presenting a balanced analysis, this review seeks to clarify the contentious roles of BCAAs in CVD, providing a foundation for future research and therapeutic strategies required because of the rising prevalence, incidence, and total burden of CVDs.
Collapse
Affiliation(s)
- Daniela Maria Tanase
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, Iasi 700111, Romania
| | - Emilia Valasciuc
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, Iasi 700111, Romania
| | - Claudia Florida Costea
- Department of Ophthalmology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- 2nd Ophthalmology Clinic, “Prof. Dr. Nicolae Oblu” Emergency Clinical Hospital, 700309 Iași, Romania
| | - Dragos Viorel Scripcariu
- Department of General Surgery, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Regional Institute of Oncology, 700483 Iasi, Romania
| | - Anca Ouatu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, Iasi 700111, Romania
| | - Loredana Liliana Hurjui
- Department of Morpho-Functional Sciences II, Physiology Discipline, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Hematology Laboratory, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Claudia Cristina Tarniceriu
- Department of Morpho-Functional Sciences I, Discipline of Anatomy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Hematology Clinic, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Diana Elena Floria
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Manuela Ciocoiu
- Department of Pathophysiology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Livia Genoveva Baroi
- Department of Surgery, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Department of Vascular Surgery, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Mariana Floria
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, Iasi 700111, Romania
| |
Collapse
|
6
|
Zhang J, Liu Z, Ni Y, Yu Y, Guo F, Lu Y, Wang X, Hao H, Li S, Wei P, Yu W, Hu W. Branched-chain amino acids promote occurrence and development of cardiovascular disease dependent on triglyceride metabolism via activation of the mTOR/SREBP-1/betatrophin pathway. Mol Cell Endocrinol 2024; 584:112164. [PMID: 38262527 DOI: 10.1016/j.mce.2024.112164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 01/25/2024]
Abstract
Branched-chain amino acid (BCAA) metabolism is associated with triglyceride (TG) metabolism and the development of cardiovascular disease (CVD). However, the underlying mechanism remains uncertain. This study included 1302 subjects and followed for 4-5 years. A hyperbranched-chain aminoacidemia rat model was induced by high fructose diet (HFTD). The relationship between BCAAs and TG level and its regulatory mechanism was investigated in vitro. As results, as baseline BCAA percentile increased, subjects had higher prevalence and incidence of T2DM, NAFLD, and CVD risk (P < 0.05). In animal model, the accumulation of BCAAs and TG and betatrophin expression were significantly elevated in the HFTD group when comparing with those in the SD group(P < 0.05). Immunofluorescence and Masson's trichrome staining revealed that the area of interstitial fibrosis was significantly increased in the HFTD group compared with control group. Met treatment significantly decreased TG levels and betatrophin expression and reversed myocardial fibrosis (P < 0.05). In vitro, LO2 cells, stimulated with 0.1-5 mM BCAAs, displayed a significant dose-dependent increase in betatrophin expression (P < 0.05). And 5 mM BCAAs stimulation significantly increased the p-mTOR and SREBP-1 expression (P < 0.05). However, this effect could be reversed by using the corresponding inhibitor or siRNAs. In conclusions, BCAAs promote occurrence and development of cardiovascular disease dependent on TG metabolism via activation of the mTOR/SREBP-1/betatrophin pathway. The study provides a new theory for the pathogenesis of CVD caused by amino acid metabolism disorders.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Endocrinology and Metabolism, Huai'an Hospital Affiliated to Xuzhou Medical University, No. 62, Huaihai South Road, Qingjiangpu District, Huai'an, 223002, China
| | - Ziyu Liu
- Department of Endocrinology and Metabolism, Huai'an Hospital Affiliated to Xuzhou Medical University, No. 62, Huaihai South Road, Qingjiangpu District, Huai'an, 223002, China
| | - Yaojun Ni
- Department of Cardiothoracic Surgery, Hospital Affiliated to Nanjing Medical College and Huai'an First People's Hospital, No. 6, Beijing West Road, Huaiyin District, Huai'an, 223021, China
| | - Yang Yu
- Department of Endocrinology and Metabolism, Huai'an Hospital Affiliated to Xuzhou Medical University, No. 62, Huaihai South Road, Qingjiangpu District, Huai'an, 223002, China
| | - Fei Guo
- Department of Endocrinology and Metabolism, Huai'an Hospital Affiliated to Xuzhou Medical University, No. 62, Huaihai South Road, Qingjiangpu District, Huai'an, 223002, China
| | - Yanwen Lu
- Department of Endocrinology and Metabolism, Huai'an Hospital Affiliated to Xuzhou Medical University, No. 62, Huaihai South Road, Qingjiangpu District, Huai'an, 223002, China
| | - Xiaoqing Wang
- Department of Endocrinology and Metabolism, Huai'an Hospital Affiliated to Xuzhou Medical University, No. 62, Huaihai South Road, Qingjiangpu District, Huai'an, 223002, China
| | - Hairong Hao
- Department of Endocrinology and Metabolism, Huai'an Hospital Affiliated to Xuzhou Medical University, No. 62, Huaihai South Road, Qingjiangpu District, Huai'an, 223002, China
| | - Shayan Li
- Department of Endocrinology and Metabolism, Huai'an Hospital Affiliated to Xuzhou Medical University, No. 62, Huaihai South Road, Qingjiangpu District, Huai'an, 223002, China
| | - Pan Wei
- Department of Endocrinology and Metabolism, Huai'an Hospital Affiliated to Xuzhou Medical University, No. 62, Huaihai South Road, Qingjiangpu District, Huai'an, 223002, China
| | - Weinan Yu
- Department of Endocrinology and Metabolism, Huai'an Hospital Affiliated to Xuzhou Medical University, No. 62, Huaihai South Road, Qingjiangpu District, Huai'an, 223002, China
| | - Wen Hu
- Department of Endocrinology and Metabolism, Huai'an Hospital Affiliated to Xuzhou Medical University, No. 62, Huaihai South Road, Qingjiangpu District, Huai'an, 223002, China.
| |
Collapse
|
7
|
Eckle T, Bertazzo J, Khatua TN, Tabatabaei SRF, Bakhtiari NM, Walker LA, Martino TA. Circadian Influences on Myocardial Ischemia-Reperfusion Injury and Heart Failure. Circ Res 2024; 134:675-694. [PMID: 38484024 PMCID: PMC10947118 DOI: 10.1161/circresaha.123.323522] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 02/08/2024] [Indexed: 03/19/2024]
Abstract
The impact of circadian rhythms on cardiovascular function and disease development is well established, with numerous studies in genetically modified animals emphasizing the circadian molecular clock's significance in the pathogenesis and pathophysiology of myocardial ischemia and heart failure progression. However, translational preclinical studies targeting the heart's circadian biology are just now emerging and are leading to the development of a novel field of medicine termed circadian medicine. In this review, we explore circadian molecular mechanisms and novel therapies, including (1) intense light, (2) small molecules modulating the circadian mechanism, and (3) chronotherapies such as cardiovascular drugs and meal timings. These promise significant clinical translation in circadian medicine for cardiovascular disease. (4) Additionally, we address the differential functioning of the circadian mechanism in males versus females, emphasizing the consideration of biological sex, gender, and aging in circadian therapies for cardiovascular disease.
Collapse
Affiliation(s)
- Tobias Eckle
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Júlia Bertazzo
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Tarak Nath Khatua
- Centre for Cardiovascular Investigations, Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Seyed Reza Fatemi Tabatabaei
- Centre for Cardiovascular Investigations, Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Naghmeh Moori Bakhtiari
- Centre for Cardiovascular Investigations, Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Lori A Walker
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Tami A. Martino
- Centre for Cardiovascular Investigations, Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
8
|
Lal H, Verma SK, Wang Y, Xie M, Young ME. Circadian Rhythms in Cardiovascular Metabolism. Circ Res 2024; 134:635-658. [PMID: 38484029 PMCID: PMC10947116 DOI: 10.1161/circresaha.123.323520] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/23/2024] [Indexed: 03/19/2024]
Abstract
Energetic demand and nutrient supply fluctuate as a function of time-of-day, in alignment with sleep-wake and fasting-feeding cycles. These daily rhythms are mirrored by 24-hour oscillations in numerous cardiovascular functional parameters, including blood pressure, heart rate, and myocardial contractility. It is, therefore, not surprising that metabolic processes also fluctuate over the course of the day, to ensure temporal needs for ATP, building blocks, and metabolism-based signaling molecules are met. What has become increasingly clear is that in addition to classic signal-response coupling (termed reactionary mechanisms), cardiovascular-relevant cells use autonomous circadian clocks to temporally orchestrate metabolic pathways in preparation for predicted stimuli/stresses (termed anticipatory mechanisms). Here, we review current knowledge regarding circadian regulation of metabolism, how metabolic rhythms are synchronized with cardiovascular function, and whether circadian misalignment/disruption of metabolic processes contribute toward the pathogenesis of cardiovascular disease.
Collapse
Affiliation(s)
- Hind Lal
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Suresh Kumar Verma
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yajing Wang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Min Xie
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Martin E. Young
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
9
|
Yeh CY, Chini LC, Davidson JW, Garcia GG, Gallagher MS, Freichels IT, Calubag MF, Rodgers AC, Green CL, Babygirija R, Sonsalla MM, Pak HH, Trautman M, Hacker TA, Miller RA, Simcox J, Lamming DW. Late-life isoleucine restriction promotes physiological and molecular signatures of healthy aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.06.527311. [PMID: 36798157 PMCID: PMC9934591 DOI: 10.1101/2023.02.06.527311] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
In defiance of the paradigm that calories from all sources are equivalent, we and others have shown that dietary protein is a dominant regulator of healthy aging. The restriction of protein or the branched-chain amino acid isoleucine promotes healthspan and extends lifespan when initiated in young or adult mice. However, many interventions are less efficacious or even deleterious when initiated in aged animals. Here, we investigate the physiological, metabolic, and molecular consequences of consuming a diet with a 67% reduction of all amino acids (Low AA), or of isoleucine alone (Low Ile), in male and female C57BL/6J.Nia mice starting at 20 months of age. We find that both diet regimens effectively reduce adiposity and improve glucose tolerance, which were benefits that were not mediated by reduced calorie intake. Both diets improve specific aspects of frailty, slow multiple molecular indicators of aging rate, and rejuvenate the aging heart and liver at the molecular level. These results demonstrate that Low AA and Low Ile diets can drive youthful physiological and molecular signatures, and support the possibility that these dietary interventions could help to promote healthy aging in older adults.
Collapse
Affiliation(s)
- Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705
| | - Lucas C.S. Chini
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705
| | - Jessica W. Davidson
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706
| | - Gonzalo G. Garcia
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109
| | - Meredith S. Gallagher
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705
| | - Isaac T. Freichels
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705
| | - Mariah F. Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705
- Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706
| | - Allison C. Rodgers
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- Cardiovascular Physiology Core Facility, University of Wisconsin-Madison, Madison, WI 53706
| | - Cara L. Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705
| | - Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI 53706
| | - Michelle M. Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705
- Comparative Biomedical Sciences Graduate Training Program, University of Wisconsin-Madison, Madison, WI 53706
| | - Heidi H. Pak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI 53706
| | - Michaela Trautman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI 53706
| | - Timothy A. Hacker
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- Cardiovascular Physiology Core Facility, University of Wisconsin-Madison, Madison, WI 53706
| | - Richard A Miller
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109
| | - Judith Simcox
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706
- Howard Hughes Medical Institute, Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Dudley W. Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705
- Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI 53706
- Comparative Biomedical Sciences Graduate Training Program, University of Wisconsin-Madison, Madison, WI 53706
| |
Collapse
|
10
|
Dial MB, Malek EM, Neblina GA, Cooper AR, Vaslieva NI, Frommer R, Girgis M, Dawn B, McGinnis GR. Effects of time-restricted exercise on activity rhythms and exercise-induced adaptations in the heart. Sci Rep 2024; 14:146. [PMID: 38168503 PMCID: PMC10761674 DOI: 10.1038/s41598-023-50113-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 12/15/2023] [Indexed: 01/05/2024] Open
Abstract
Circadian rhythms play a crucial role in the regulation of various physiological processes, including cardiovascular function and metabolism. Exercise provokes numerous beneficial adaptations in heart, including physiological hypertrophy, and serves to shift circadian rhythms. This study investigated the impact of time-restricted exercise training on exercise-induced adaptations in the heart and locomotor activity rhythms. Male mice (n = 45) were allocated to perform voluntary, time-restricted exercise in the early active phase (EAP), late active phase (LAP), or remain sedentary (SED) for 6 weeks. Subsequently, mice were allowed 24-h ad libitum access to the running wheel to assess diurnal rhythms in locomotor activity. Heart weight and cross-sectional area were measured at sacrifice, and cardiac protein and gene expression levels were assessed for markers of mitochondrial abundance and circadian clock gene expression. Mice rapidly adapted to wheel running, with EAP mice exhibiting a significantly greater running distance compared to LAP mice. Time-restricted exercise induced a shift in voluntary wheel activity during the 24-h free access period, with the acrophase in activity being significantly earlier in EAP mice compared to LAP mice. Gene expression analysis revealed a higher expression of Per1 in LAP mice. EAP exercise elicited greater cardiac hypertrophy compared to LAP exercise. These findings suggest that the timing of exercise affects myocardial adaptations, with exercise in the early active phase inducing hypertrophy in the heart. Understanding the time-of-day dependent response to exercise in the heart may have implications for optimizing exercise interventions for cardiovascular health.
Collapse
Affiliation(s)
- Michael B Dial
- Department of Kinesiology and Nutrition Sciences, University of Nevada, Las Vegas, 4505 S. Maryland Parkway, Bigelow Health Sciences (BHS) Building 323, Las Vegas, NV, 89154, USA
| | - Elias M Malek
- Department of Kinesiology and Nutrition Sciences, University of Nevada, Las Vegas, 4505 S. Maryland Parkway, Bigelow Health Sciences (BHS) Building 323, Las Vegas, NV, 89154, USA
| | - Greco A Neblina
- Department of Kinesiology and Nutrition Sciences, University of Nevada, Las Vegas, 4505 S. Maryland Parkway, Bigelow Health Sciences (BHS) Building 323, Las Vegas, NV, 89154, USA
| | - Austin R Cooper
- Department of Kinesiology and Nutrition Sciences, University of Nevada, Las Vegas, 4505 S. Maryland Parkway, Bigelow Health Sciences (BHS) Building 323, Las Vegas, NV, 89154, USA
| | - Nikoleta I Vaslieva
- Department of Kinesiology and Nutrition Sciences, University of Nevada, Las Vegas, 4505 S. Maryland Parkway, Bigelow Health Sciences (BHS) Building 323, Las Vegas, NV, 89154, USA
| | - Rebecca Frommer
- Department of Internal Medicine, Kirk Kerkorian School of Medicine, University of Nevada, Las Vegas, Las Vegas, NV, USA
| | - Magdy Girgis
- Department of Internal Medicine, Kirk Kerkorian School of Medicine, University of Nevada, Las Vegas, Las Vegas, NV, USA
| | - Buddhadeb Dawn
- Department of Internal Medicine, Kirk Kerkorian School of Medicine, University of Nevada, Las Vegas, Las Vegas, NV, USA
| | - Graham R McGinnis
- Department of Kinesiology and Nutrition Sciences, University of Nevada, Las Vegas, 4505 S. Maryland Parkway, Bigelow Health Sciences (BHS) Building 323, Las Vegas, NV, 89154, USA.
| |
Collapse
|
11
|
Young ME. The Cardiac Circadian Clock: Implications for Cardiovascular Disease and its Treatment. JACC Basic Transl Sci 2023; 8:1613-1628. [PMID: 38205356 PMCID: PMC10774593 DOI: 10.1016/j.jacbts.2023.03.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/23/2023] [Accepted: 03/23/2023] [Indexed: 01/12/2024]
Abstract
Virtually all aspects of physiology fluctuate with respect to the time of day. This is beautifully exemplified by cardiovascular physiology, for which blood pressure and electrophysiology exhibit robust diurnal oscillations. At molecular/biochemical levels (eg, transcription, translation, signaling, metabolism), cardiovascular-relevant tissues (such as the heart) are profoundly different during the day vs the night. Unfortunately, this in turn contributes toward 24-hour rhythms in both risk of adverse event onset (eg, arrhythmias, myocardial infarction) and pathogenesis severity (eg, extent of ischemic damage). Accumulating evidence indicates that cell-autonomous timekeeping mechanisms, termed circadian clocks, temporally govern biological processes known to play critical roles in cardiovascular function/dysfunction. In this paper, a comprehensive review of our current understanding of the cardiomyocyte circadian clock during both health and disease is detailed. Unprecedented basic, translational, and epidemiologic studies support a need to implement chronobiological considerations in strategies designed for both prevention and treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Martin E. Young
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
12
|
Latimer MN, Williams LJ, Shanmugan G, Carpenter BJ, Lazar MA, Dierickx P, Young ME. Cardiomyocyte-specific disruption of the circadian BMAL1-REV-ERBα/β regulatory network impacts distinct miRNA species in the murine heart. Commun Biol 2023; 6:1149. [PMID: 37952007 PMCID: PMC10640639 DOI: 10.1038/s42003-023-05537-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 11/01/2023] [Indexed: 11/14/2023] Open
Abstract
Circadian disruption increases cardiovascular disease (CVD) risk, through poorly understood mechanisms. Given that small RNA species are critical modulators of cardiac physiology/pathology, we sought to determine the extent to which cardiomyocyte circadian clock (CCC) disruption impacts cardiac small RNA species. Accordingly, we collected hearts from cardiomyocyte-specific Bmal1 knockout (CBK; a model of CCC disruption) and littermate control (CON) mice at multiple times of the day, followed by small RNA-seq. The data reveal 47 differentially expressed miRNAs species in CBK hearts. Subsequent bioinformatic analyses predict that differentially expressed miRNA species in CBK hearts influence processes such as circadian rhythmicity, cellular signaling, and metabolism. Of the induced miRNAs in CBK hearts, 7 are predicted to be targeted by the transcriptional repressors REV-ERBα/β (integral circadian clock components that are directly regulated by BMAL1). Similar to CBK hearts, cardiomyocyte-specific Rev-erbα/β double knockout (CM-RevDKO) mouse hearts exhibit increased let-7c-1-3p, miR-23b-5p, miR-139-3p, miR-5123, and miR-7068-3p levels. Importantly, 19 putative targets of these 5 miRNAs are commonly repressed in CBK and CM-RevDKO heart (of which 16 are targeted by let-7c-1-3p). These observations suggest that disruption of the circadian BMAL1-REV-ERBα/β regulatory network in the heart induces distinct miRNAs, whose mRNA targets impact critical cellular functions.
Collapse
Affiliation(s)
- Mary N Latimer
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lamario J Williams
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gobinath Shanmugan
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Bryce J Carpenter
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Mitchell A Lazar
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Pieterjan Dierickx
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
| | - Martin E Young
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
13
|
Schulman-Geltzer EB, Collins HE, Hill BG, Fulghum KL. Coordinated Metabolic Responses Facilitate Cardiac Growth in Pregnancy and Exercise. Curr Heart Fail Rep 2023; 20:441-450. [PMID: 37581772 PMCID: PMC10589193 DOI: 10.1007/s11897-023-00622-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/19/2023] [Indexed: 08/16/2023]
Abstract
PURPOSE OF REVIEW Pregnancy and exercise are systemic stressors that promote physiological growth of the heart in response to repetitive volume overload and maintenance of cardiac output. This type of remodeling is distinct from pathological hypertrophy and involves different metabolic mechanisms that facilitate growth; however, it remains unclear how metabolic changes in the heart facilitate growth and if these processes are similar in both pregnancy- and exercise-induced cardiac growth. RECENT FINDINGS The ability of the heart to metabolize a myriad of substrates balances cardiac demands for energy provision and anabolism. During pregnancy, coordination of hormonal status with cardiac reductions in glucose oxidation appears important for physiological growth. During exercise, a reduction in cardiac glucose oxidation also appears important for physiological growth, which could facilitate shuttling of glucose-derived carbons into biosynthetic pathways for growth. Understanding the metabolic underpinnings of physiological cardiac growth could provide insight to optimize cardiovascular health and prevent deleterious remodeling, such as that which occurs from postpartum cardiomyopathy and heart failure. This short review highlights the metabolic mechanisms known to facilitate pregnancy-induced and exercise-induced cardiac growth, both of which require changes in cardiac glucose metabolism for the promotion of growth. In addition, we mention important similarities and differences of physiological cardiac growth in these models as well as discuss current limitations in our understanding of metabolic changes that facilitate growth.
Collapse
Affiliation(s)
- Emily B Schulman-Geltzer
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Helen E Collins
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Bradford G Hill
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Kyle L Fulghum
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Department of Medicine, University of Louisville, Louisville, KY, USA.
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
14
|
Mia S, Sonkar R, Williams L, Latimer MN, Rawnsley DR, Rana S, He J, Dierickx P, Kim T, Xie M, Habegger KM, Kubo M, Zhou L, Thomsen MB, Prabhu SD, Frank SJ, Brookes PS, Lazar MA, Diwan A, Young ME. Novel Roles for the Transcriptional Repressor E4BP4 in Both Cardiac Physiology and Pathophysiology. JACC Basic Transl Sci 2023; 8:1141-1156. [PMID: 37791313 PMCID: PMC10543917 DOI: 10.1016/j.jacbts.2023.03.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 10/05/2023]
Abstract
Circadian clocks temporally orchestrate biological processes critical for cellular/organ function. For example, the cardiomyocyte circadian clock modulates cardiac metabolism, signaling, and electrophysiology over the course of the day, such that, disruption of the clock leads to age-onset cardiomyopathy (through unknown mechanisms). Here, we report that genetic disruption of the cardiomyocyte clock results in chronic induction of the transcriptional repressor E4BP4. Importantly, E4BP4 deletion prevents age-onset cardiomyopathy following clock disruption. These studies also indicate that E4BP4 regulates both cardiac metabolism (eg, fatty acid oxidation) and electrophysiology (eg, QT interval). Collectively, these studies reveal that E4BP4 is a novel regulator of both cardiac physiology and pathophysiology.
Collapse
Affiliation(s)
- Sobuj Mia
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ravi Sonkar
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Lamario Williams
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mary N. Latimer
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - David R. Rawnsley
- Departments of Medicine, Cell Biology and Physiology, Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Samir Rana
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jin He
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Pieterjan Dierickx
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Teayoun Kim
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Min Xie
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kirk M. Habegger
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Masato Kubo
- Research Institute for Biomedical Science, Tokyo University of Science, Chiba, Japan
- Laboratory for Cytokine Regulation, RIKEN Center for Integrative Medical Sciences (IMS), RIKEN Yokohama Institute, Kanagawa, Japan
| | - Lufang Zhou
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Morten B. Thomsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Demark
| | - Sumanth D. Prabhu
- Departments of Medicine, Cell Biology and Physiology, Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Stuart J. Frank
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Endocrinology Section, Birmingham VAMC Medical Service, Birmingham, Alabama, USA
| | - Paul S. Brookes
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, New York, USA
| | - Mitchell A. Lazar
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Abhinav Diwan
- Departments of Medicine, Cell Biology and Physiology, Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri, USA
- John Cochran VA Medical Center, St. Louis, Missouri, USA
| | - Martin E. Young
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
15
|
Roth Flach RJ, Bollinger E, Reyes AR, Laforest B, Kormos BL, Liu S, Reese MR, Martinez Alsina LA, Buzon L, Zhang Y, Bechle B, Rosado A, Sahasrabudhe PV, Knafels J, Bhattacharya SK, Omoto K, Stansfield JC, Hurley LD, Song L, Luo L, Breitkopf SB, Monetti M, Cunio T, Tierney B, Geoly FJ, Delmore J, Siddall CP, Xue L, Yip KN, Kalgutkar AS, Miller RA, Zhang BB, Filipski KJ. Small molecule branched-chain ketoacid dehydrogenase kinase (BDK) inhibitors with opposing effects on BDK protein levels. Nat Commun 2023; 14:4812. [PMID: 37558654 PMCID: PMC10412597 DOI: 10.1038/s41467-023-40536-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 07/25/2023] [Indexed: 08/11/2023] Open
Abstract
Branched chain amino acid (BCAA) catabolic impairments have been implicated in several diseases. Branched chain ketoacid dehydrogenase (BCKDH) controls the rate limiting step in BCAA degradation, the activity of which is inhibited by BCKDH kinase (BDK)-mediated phosphorylation. Screening efforts to discover BDK inhibitors led to identification of thiophene PF-07208254, which improved cardiometabolic endpoints in mice. Structure-activity relationship studies led to identification of a thiazole series of BDK inhibitors; however, these inhibitors did not improve metabolism in mice upon chronic administration. While the thiophenes demonstrated sustained branched chain ketoacid (BCKA) lowering and reduced BDK protein levels, the thiazoles increased BCKAs and BDK protein levels. Thiazoles increased BDK proximity to BCKDH-E2, whereas thiophenes reduced BDK proximity to BCKDH-E2, which may promote BDK degradation. Thus, we describe two BDK inhibitor series that possess differing attributes regarding BDK degradation or stabilization and provide a mechanistic understanding of the desirable features of an effective BDK inhibitor.
Collapse
Affiliation(s)
- Rachel J Roth Flach
- Pfizer Worldwide Research, Development & Medical, Cambridge, MA, 02139, USA.
| | - Eliza Bollinger
- Pfizer Worldwide Research, Development & Medical, Cambridge, MA, 02139, USA
| | - Allan R Reyes
- Pfizer Worldwide Research, Development & Medical, Cambridge, MA, 02139, USA
| | - Brigitte Laforest
- Pfizer Worldwide Research, Development & Medical, Cambridge, MA, 02139, USA
| | - Bethany L Kormos
- Pfizer Worldwide Research, Development & Medical, Cambridge, MA, 02139, USA
| | - Shenping Liu
- Pfizer Worldwide Research, Development & Medical, Groton, CT, 06340, USA
| | - Matthew R Reese
- Pfizer Worldwide Research, Development & Medical, Groton, CT, 06340, USA
| | | | - Leanne Buzon
- Pfizer Worldwide Research, Development & Medical, Groton, CT, 06340, USA
| | - Yuan Zhang
- Pfizer Worldwide Research, Development & Medical, Groton, CT, 06340, USA
| | - Bruce Bechle
- Pfizer Worldwide Research, Development & Medical, Groton, CT, 06340, USA
| | - Amy Rosado
- Pfizer Worldwide Research, Development & Medical, Groton, CT, 06340, USA
| | | | - John Knafels
- Pfizer Worldwide Research, Development & Medical, Groton, CT, 06340, USA
| | | | - Kiyoyuki Omoto
- Pfizer Worldwide Research, Development & Medical, Cambridge, MA, 02139, USA
| | - John C Stansfield
- Pfizer Worldwide Research, Development & Medical, Cambridge, MA, 02139, USA
| | - Liam D Hurley
- Pfizer Worldwide Research, Development & Medical, Cambridge, MA, 02139, USA
| | - LouJin Song
- Pfizer Worldwide Research, Development & Medical, Cambridge, MA, 02139, USA
| | - Lina Luo
- Pfizer Worldwide Research, Development & Medical, Groton, CT, 06340, USA
| | | | - Mara Monetti
- Pfizer Worldwide Research, Development & Medical, Cambridge, MA, 02139, USA
| | - Teresa Cunio
- Pfizer Worldwide Research, Development & Medical, Cambridge, MA, 02139, USA
| | - Brendan Tierney
- Pfizer Worldwide Research, Development & Medical, Groton, CT, 06340, USA
| | - Frank J Geoly
- Pfizer Worldwide Research, Development & Medical, Groton, CT, 06340, USA
| | - Jake Delmore
- Pfizer Worldwide Research, Development & Medical, Cambridge, MA, 02139, USA
| | - C Parker Siddall
- Pfizer Worldwide Research, Development & Medical, Cambridge, MA, 02139, USA
| | - Liang Xue
- Pfizer Worldwide Research, Development & Medical, Cambridge, MA, 02139, USA
| | - Ka N Yip
- Pfizer Worldwide Research, Development & Medical, Cambridge, MA, 02139, USA
| | - Amit S Kalgutkar
- Pfizer Worldwide Research, Development & Medical, Cambridge, MA, 02139, USA
| | - Russell A Miller
- Pfizer Worldwide Research, Development & Medical, Cambridge, MA, 02139, USA
| | - Bei B Zhang
- Pfizer Worldwide Research, Development & Medical, Cambridge, MA, 02139, USA
| | - Kevin J Filipski
- Pfizer Worldwide Research, Development & Medical, Cambridge, MA, 02139, USA.
| |
Collapse
|
16
|
Mihaylova MM, Chaix A, Delibegovic M, Ramsey JJ, Bass J, Melkani G, Singh R, Chen Z, Ja WW, Shirasu-Hiza M, Latimer MN, Mattison JA, Thalacker-Mercer AE, Dixit VD, Panda S, Lamming DW. When a calorie is not just a calorie: Diet quality and timing as mediators of metabolism and healthy aging. Cell Metab 2023; 35:1114-1131. [PMID: 37392742 PMCID: PMC10528391 DOI: 10.1016/j.cmet.2023.06.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/07/2023] [Accepted: 06/13/2023] [Indexed: 07/03/2023]
Abstract
An epidemic of obesity has affected large portions of the world, increasing the risk of developing many different age-associated diseases, including cancer, cardiovascular disease, and diabetes. In contrast with the prevailing notion that "a calorie is just a calorie," there are clear differences, within and between individuals, in the metabolic response to different macronutrient sources. Recent findings challenge this oversimplification; calories from different macronutrient sources or consumed at different times of day have metabolic effects beyond their value as fuel. Here, we summarize discussions conducted at a recent NIH workshop that brought together experts in calorie restriction, macronutrient composition, and time-restricted feeding to discuss how dietary composition and feeding schedule impact whole-body metabolism, longevity, and healthspan. These discussions may provide insights into the long-sought molecular mechanisms engaged by calorie restriction to extend lifespan, lead to novel therapies, and potentially inform the development of a personalized food-as-medicine approach to healthy aging.
Collapse
Affiliation(s)
- Maria M Mihaylova
- Department of Biological Chemistry and Pharmacology, College of Medicine, The Ohio State University, Columbus, OH, USA; The Ohio State University, Comprehensive Cancer Center, Wexner Medical Center, Arthur G. James Cancer Hospital, Columbus, OH, USA.
| | - Amandine Chaix
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
| | - Mirela Delibegovic
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill Health Campus, Aberdeen, UK
| | - Jon J Ramsey
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, USA
| | - Joseph Bass
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Girish Melkani
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Rajat Singh
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Zheng Chen
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - William W Ja
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Michele Shirasu-Hiza
- Department of Genetics and Development, Columbia University Medical Center, New York, NY, USA
| | - Mary N Latimer
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Julie A Mattison
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Anna E Thalacker-Mercer
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Vishwa Deep Dixit
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA; Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA; Yale Center for Research on Aging, Yale School of Medicine, New Haven, CT, USA
| | - Satchidananda Panda
- Regulatory Biology Lab, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
| |
Collapse
|
17
|
Paula ABR, Resende LT, Jardim IABA, Portes AMO, Isoldi MC. The role of environmental signals in the expression of rhythmic cardiac proteins and their influence on cardiac pathologies. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 137:205-223. [PMID: 37709377 DOI: 10.1016/bs.apcsb.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
We know that numerous proteins expressed in the heart are influenced by environmental signals (such as light and diet), which cause either an increase or decrease in their expression. Cardiovascular health is sensitive to diet composition (macronutrient content), as well as the percentage of energy, frequency and regularity of meal intake during the 24-hour cycle, and the fasting period. Furthermore, light is an important synchronizer of the circadian clock and, in turn, of several physiological processes, among them cardiovascular physiology. In this chapter, we address the effects of these environmental cues and the known mechanisms that lead to this variation in protein expression in the heart, as well as cardiac function.
Collapse
Affiliation(s)
- Ana Beatriz Rezende Paula
- Laboratory of Cell Signaling, Research Center in Biological Science, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Bauxita, Ouro Preto, Brazil.
| | - Letícia Teresinha Resende
- Laboratory of Cell Signaling, Research Center in Biological Science, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Bauxita, Ouro Preto, Brazil
| | - Isabela Alcântara Barretto Araújo Jardim
- Laboratory of Cell Signaling, Research Center in Biological Science, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Bauxita, Ouro Preto, Brazil
| | - Alexandre Martins Oliveira Portes
- Laboratory of Cell Signaling, Research Center in Biological Science, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Bauxita, Ouro Preto, Brazil
| | - Mauro César Isoldi
- Laboratory of Cell Signaling, Research Center in Biological Science, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Bauxita, Ouro Preto, Brazil
| |
Collapse
|
18
|
Krishnamurthy HK, Reddy S, Jayaraman V, Krishna K, Song Q, Wang T, Bei K, Rajasekaran JJ. Preliminary Study on the Association of Serum Branched-Chain Amino Acids With Lipid and Hepatic Markers. Cardiol Res 2023; 14:167-175. [PMID: 37304913 PMCID: PMC10257501 DOI: 10.14740/cr1454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/27/2023] [Indexed: 06/13/2023] Open
Abstract
Background Serum levels of branched-chain amino acids (BCAAs) are associated with various vital physiological functions and thus elevation in circulating levels results in several metabolic disturbances. Serum levels of BCAAs are strong predictors of various metabolic disorders. Their association with cardiovascular health is uncertain. The study aimed to investigate the association of BCAAs with circulating levels of vital cardiovascular and hepatic markers. Methods The study population of 714 individuals was included from the population tested for the vital cardio and hepatic biomarkers at the Vibrant America Clinical Laboratories. The subjects were stratified into four quartiles based on the serum levels of BCAAs, and their association with vital markers was studied using the Kruskal-Wallis test. Pearson's correlation analyzed the univariant relationship of BCAAs with selected cardio and hepatic markers. Results BCAAs exhibited a strong negative correlation with serum HDL. Serum triglycerides were found to have a positive correlation with serum levels of leucine and valine. Univariant analysis exhibited a strong negative correlation between serum levels of BCAAs and HDL, and a positive correlation was observed between triglycerides and amino acids isoleucine and leucine. Among analyzed hepatic markers, alanine transaminase exhibited a considerable association with BCAAs. Conclusions The elevated levels of serum BCAAs are strongly associated with serum HDL and triglycerides. Consumption of these supplements must be in coordination with healthcare providers to avoid metabolic and cardiovascular risk.
Collapse
Affiliation(s)
| | | | | | | | - Qi Song
- Vibrant America LLC., San Carlos, CA, USA
| | - Tianhao Wang
- Vibrant Sciences LLC., San Carlos, CA 94070, USA
| | - Kang Bei
- Vibrant Sciences LLC., San Carlos, CA 94070, USA
| | | |
Collapse
|
19
|
Interplay between Exercise, Circadian Rhythm, and Cardiac Metabolism and Remodeling. CURRENT OPINION IN PHYSIOLOGY 2023. [DOI: 10.1016/j.cophys.2023.100643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
20
|
Abstract
Research conducted in the past 15 years has yielded crucial insights that are reshaping our understanding of the systems physiology of branched-chain amino acid (BCAA) metabolism and the molecular mechanisms underlying the close relationship between BCAA homeostasis and cardiovascular health. The rapidly evolving literature paints a complex picture, in which numerous tissue-specific and disease-specific modes of BCAA regulation initiate a diverse set of molecular mechanisms that connect changes in BCAA homeostasis to the pathogenesis of cardiovascular diseases, including myocardial infarction, ischaemia-reperfusion injury, atherosclerosis, hypertension and heart failure. In this Review, we outline the current understanding of the major factors regulating BCAA abundance and metabolic fate, highlight molecular mechanisms connecting impaired BCAA homeostasis to cardiovascular disease, discuss the epidemiological evidence connecting BCAAs with various cardiovascular disease states and identify current knowledge gaps requiring further investigation.
Collapse
Affiliation(s)
- Robert W McGarrah
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA.
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC, USA.
- Department of Medicine, Division of Cardiology, Duke University, Durham, NC, USA.
| | - Phillip J White
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA.
- Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Duke University, Durham, NC, USA.
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA.
| |
Collapse
|
21
|
Abstract
Driven by autonomous molecular clocks that are synchronized by a master pacemaker in the suprachiasmatic nucleus, cardiac physiology fluctuates in diurnal rhythms that can be partly or entirely circadian. Cardiac contractility, metabolism, and electrophysiology, all have diurnal rhythms, as does the neurohumoral control of cardiac and kidney function. In this review, we discuss the evidence that circadian biology regulates cardiac function, how molecular clocks may relate to the pathogenesis of heart failure, and how chronotherapeutics might be applied in heart failure. Disrupting molecular clocks can lead to heart failure in animal models, and the myocardial response to injury seems to be conditioned by the time of day. Human studies are consistent with these findings, and they implicate the clock and circadian rhythms in the pathogenesis of heart failure. Certain circadian rhythms are maintained in patients with heart failure, a factor that can guide optimal timing of therapy. Pharmacologic and nonpharmacologic manipulation of circadian rhythms and molecular clocks show promise in the prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Nadim El Jamal
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ronan Lordan
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sarah L. Teegarden
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Tilo Grosser
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Translational Pharmacology, Bielefeld University, Bielefeld, Germany
| | - Garret FitzGerald
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
22
|
The Effect of Diet on the Cardiac Circadian Clock in Mice: A Systematic Review. Metabolites 2022; 12:metabo12121273. [PMID: 36557311 PMCID: PMC9786298 DOI: 10.3390/metabo12121273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/19/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022] Open
Abstract
Circadian rhythms play important roles in regulating physiological and behavioral processes. These are adjusted by environmental cues, such as diet, which acts by synchronizing or attenuating the circadian rhythms of peripheral clocks, such as the liver, intestine, pancreas, white and brown adipose tissue, lungs, kidneys, as well as the heart. Some studies point to the influence of diet composition, feeding timing, and dietary restriction on metabolic homeostasis and circadian rhythms at various levels. Therefore, this systematic review aimed to discuss studies addressing the effect of diet on the heart clock in animal models and, additionally, the chronodisruption of the clock and its relation to the development of cardiovascular disorders in the last 15 years. A search was conducted in the PubMed, Scopus, and Embase databases. The PRISMA guide was used to construct the article. Nineteen studies met all inclusion and exclusion criteria. In summary, these studies have linked the circadian clock to cardiovascular health and suggested that maintaining a robust circadian system may reduce the risks of cardiometabolic and cardiovascular diseases. The effect of time-of-day-dependent eating on the modulation of circadian rhythms of the cardiac clock and energy homeostasis is notable, among its deleterious effects predominantly in the sleep (light) phase and/or at the end of the active phase.
Collapse
|
23
|
Identifying a metabolomics profile associated with masked hypertension in two independent cohorts: Data from the African-PREDICT and SABPA studies. Hypertens Res 2022; 45:1781-1793. [PMID: 36056205 DOI: 10.1038/s41440-022-01010-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/27/2022] [Accepted: 07/21/2022] [Indexed: 12/15/2022]
Abstract
Individuals with masked hypertension (MHT) have a greater risk of adverse cardiovascular outcomes than normotensive (NT) individuals. Exploring metabolomic differences between NT and MHT individuals may help provide a better understanding of the etiology of MHT. We analyzed data from 910 young participants (83% NT and 17% MHT) (mean age 24 ± 3 years) from the African-PREDICT and 210 older participants (63% NT and 37% MHT) from the SABPA (mean age 42 ± 9.6 years) studies. Clinic and ambulatory blood pressures (BPs) were used to define BP phenotypes. Urinary amino acids and acylcarnitines were measured using liquid chromatography time-of-flight mass spectrometry in SABPA and liquid chromatography tandem mass spectrometry in the African-PREDICT studies. In the SABPA study, amino acids (leucine/isoleucine, valine, methionine, phenylalanine), free carnitine (C0-carnitine), and acylcarnitines C3 (propionyl)-, C4 (butyryl)-carnitine and total acylcarnitine) were higher in MHT than NT adults. In the African-PREDICT study, C0- and C5-carnitines were higher in MHT individuals. With unadjusted analyses in NT adults from the SABPA study, ambulatory SBP correlated positively with only C3-carnitine. In MHT individuals, positive correlations of ambulatory SBP with leucine/isoleucine, valine, methionine, phenylalanine, C0-carnitine and C3-carnitine were evident (all p < 0.05). In the African-PREDICT study, ambulatory SBP correlated positively with C0-carnitine (r = 0.101; p = 0.006) and C5-carnitine (r = 0.195; p < 0.001) in NT adults and C5-carnitine in MHT individuals (r = 0.169; p = 0.034). We demonstrated differences between the metabolomic profiles of NT and MHT adults, which may reflect different stages in the alteration of branched-chain amino acid metabolism early on and later in life.
Collapse
|
24
|
Bollinger E, Peloquin M, Libera J, Albuquerque B, Pashos E, Shipstone A, Hadjipanayis A, Sun Z, Xing G, Clasquin M, Stansfield JC, Tierney B, Gernhardt S, Siddall CP, Greizer T, Geoly FJ, Vargas SR, Gao LC, Williams G, Marshall M, Rosado A, Steppan C, Filipski KJ, Zhang BB, Miller RA, Roth Flach RJ. BDK inhibition acts as a catabolic switch to mimic fasting and improve metabolism in mice. Mol Metab 2022; 66:101611. [PMID: 36220546 PMCID: PMC9589198 DOI: 10.1016/j.molmet.2022.101611] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/05/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE Branched chain amino acid (BCAA) catabolic defects are implicated to be causal determinates of multiple diseases. This work aimed to better understand how enhancing BCAA catabolism affected metabolic homeostasis as well as the mechanisms underlying these improvements. METHODS The rate limiting step of BCAA catabolism is the irreversible decarboxylation by the branched chain ketoacid dehydrogenase (BCKDH) enzyme complex, which is post-translationally controlled through phosphorylation by BCKDH kinase (BDK). This study utilized BT2, a small molecule allosteric inhibitor of BDK, in multiple mouse models of metabolic dysfunction and NAFLD including the high fat diet (HFD) model with acute and chronic treatment paradigms, the choline deficient and methionine minimal high fat diet (CDAHFD) model, and the low-density lipoprotein receptor null mouse model (Ldlr-/-). shRNA was additionally used to knock down BDK in liver to elucidate liver-specific effects of BDK inhibition in HFD-fed mice. RESULTS A rapid improvement in insulin sensitivity was observed in HFD-fed and lean mice after BT2 treatment. Resistance to steatosis was assessed in HFD-fed mice, CDAHFD-fed mice, and Ldlr-/- mice. In all cases, BT2 treatment reduced steatosis and/or inflammation. Fasting and refeeding demonstrated a lack of response to feeding-induced changes in plasma metabolites including insulin and beta-hydroxybutyrate and hepatic gene changes in BT2-treated mice. Mechanistically, BT2 treatment acutely altered the expression of genes involved in fatty acid oxidation and lipogenesis in liver, and upstream regulator analysis suggested that BT2 treatment activated PPARα. However, BT2 did not directly activate PPARα in vitro. Conversely, shRNA-AAV-mediated knockdown of BDK specifically in liver in vivo did not demonstrate any effects on glycemia, steatosis, or PPARα-mediated gene expression in mice. CONCLUSIONS These data suggest that BT2 treatment acutely improves metabolism and liver steatosis in multiple mouse models. While many molecular changes occur in liver in BT2-treated mice, these changes were not observed in mice with AAV-mediated shRNA knockdown of BDK. All together, these data suggest that systemic BDK inhibition is required to improve metabolism and steatosis by prolonging a fasting signature in a paracrine manner. Therefore, BCAA may act as a "fed signal" to promote nutrient storage and reduced systemic BCAA levels as shown in this study via BDK inhibition may act as a "fasting signal" to prolong the catabolic state.
Collapse
Affiliation(s)
- Eliza Bollinger
- Internal Medicine Research Unit, Pfizer Inc, Cambridge MA 02139, USA
| | - Matthew Peloquin
- Internal Medicine Research Unit, Pfizer Inc, Cambridge MA 02139, USA
| | - Jenna Libera
- Internal Medicine Research Unit, Pfizer Inc, Cambridge MA 02139, USA
| | - Bina Albuquerque
- Internal Medicine Research Unit, Pfizer Inc, Cambridge MA 02139, USA
| | - Evanthia Pashos
- Internal Medicine Research Unit, Pfizer Inc, Cambridge MA 02139, USA
| | - Arun Shipstone
- Inflammation & Immunology Research Unit, Pfizer Inc, Cambridge MA 02139, USA
| | - Angela Hadjipanayis
- Inflammation & Immunology Research Unit, Pfizer Inc, Cambridge MA 02139, USA
| | - Zhongyuan Sun
- Internal Medicine Research Unit, Pfizer Inc, Cambridge MA 02139, USA
| | - Gang Xing
- Internal Medicine Research Unit, Pfizer Inc, Cambridge MA 02139, USA
| | - Michelle Clasquin
- Internal Medicine Research Unit, Pfizer Inc, Cambridge MA 02139, USA
| | | | | | | | - C. Parker Siddall
- Internal Medicine Research Unit, Pfizer Inc, Cambridge MA 02139, USA
| | - Timothy Greizer
- Internal Medicine Research Unit, Pfizer Inc, Cambridge MA 02139, USA
| | - Frank J. Geoly
- Drug Safety Research and Development, Pfizer Inc, Groton CT 06340, USA
| | - Sarah R. Vargas
- Drug Safety Research and Development, Pfizer Inc, Groton CT 06340, USA
| | - Lily C. Gao
- Internal Medicine Research Unit, Pfizer Inc, Cambridge MA 02139, USA
| | - George Williams
- Internal Medicine Research Unit, Pfizer Inc, Cambridge MA 02139, USA
| | | | - Amy Rosado
- Medicine Design, Pfizer Inc, Groton, CT 06340, USA
| | | | | | - Bei B. Zhang
- Internal Medicine Research Unit, Pfizer Inc, Cambridge MA 02139, USA
| | - Russell A. Miller
- Internal Medicine Research Unit, Pfizer Inc, Cambridge MA 02139, USA
| | - Rachel J. Roth Flach
- Internal Medicine Research Unit, Pfizer Inc, Cambridge MA 02139, USA,Corresponding author. Pfizer Inc, 1 Portland St, Cambridge MA 02139, USA.
| |
Collapse
|
25
|
Metabolomic Profiling of End-Stage Heart Failure Secondary to Chronic Chagas Cardiomyopathy. Int J Mol Sci 2022; 23:ijms231810456. [PMID: 36142367 PMCID: PMC9499603 DOI: 10.3390/ijms231810456] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/25/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
Chronic Chagas cardiomyopathy (CCC) is the most frequent and severe clinical form of chronic Chagas disease, representing one of the leading causes of morbidity and mortality in Latin America, and a growing global public health problem. There is currently no approved treatment for CCC; however, omics technologies have enabled significant progress to be made in the search for new therapeutic targets. The metabolic alterations associated with pathogenic mechanisms of CCC and their relationship to cellular and immunopathogenic processes in cardiac tissue remain largely unknown. This exploratory study aimed to evaluate the potential underlying pathogenic mechanisms in the failing myocardium of patients with end-stage heart failure (ESHF) secondary to CCC by applying an untargeted metabolomic profiling approach. Cardiac tissue samples from the left ventricle of patients with ESHF of CCC etiology (n = 7) and healthy donors (n = 7) were analyzed using liquid chromatography-mass spectrometry. Metabolite profiles showed altered branched-chain amino acid and acylcarnitine levels, decreased fatty acid uptake and oxidation, increased activity of the pentose phosphate pathway, dysregulation of the TCA cycle, and alterations in critical cellular antioxidant systems. These findings suggest processes of energy deficit, alterations in substrate availability, and enhanced production of reactive oxygen species in the affected myocardium. This profile potentially contributes to the development and maintenance of a chronic inflammatory state that leads to progression and severity of CCC. Further studies involving larger sample sizes and comparisons with heart failure patients without CCC are needed to validate these results, opening an avenue to investigate new therapeutic approaches for the treatment and prevention of progression of this unique and severe cardiomyopathy.
Collapse
|
26
|
Yong YN, Henry CJ, Haldar S. Is There a Utility of Chrono-Specific Diets in Improving Cardiometabolic Health? Mol Nutr Food Res 2022; 66:e2200043. [PMID: 35856629 DOI: 10.1002/mnfr.202200043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 05/31/2022] [Indexed: 11/10/2022]
Abstract
Modern lifestyle is generally associated with the consumption of three main meals per day, one of which is typically in the evening or at night. It is also well established that consumption of meals in the later part of the day, notably in the evenings, is associated with circadian desynchrony, which in turn increases the risk of non-communicable diseases, particularly cardiometabolic diseases. While it is not feasible to avoid food consumption during the evenings altogether, there is an opportunity to provide chrono-specific, diet-based solutions to mitigate some of these risks. To date, there has been substantial progress in the understanding of chrononutrition, with evidence derived mainly from in vitro and in vivo animal studies. Some of these approaches include the manipulation of the quality and quantity of certain nutrients to be consumed at specific times of the day, as well as incorporating certain dietary components (macronutrients, micronutrients, or non-nutrient bioactives, including polyphenols) with the ability to modulate circadian rhythmicity. However, robust human studies are generally lacking. In this review, the study has consolidated and critically appraised the current evidence base, with an aim to translate these findings to improve cardiometabolic health and provides recommendations to move this field forward.
Collapse
Affiliation(s)
- Yi Ning Yong
- Clinical Nutrition Research Centre, Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), Singapore, 117599, Singapore
| | - Christiani Jeyakumar Henry
- Clinical Nutrition Research Centre, Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), Singapore, 117599, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, 117599, Singapore
| | - Sumanto Haldar
- Clinical Nutrition Research Centre, Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), Singapore, 117599, Singapore
| |
Collapse
|
27
|
Trautman ME, Richardson NE, Lamming DW. Protein restriction and branched-chain amino acid restriction promote geroprotective shifts in metabolism. Aging Cell 2022; 21:e13626. [PMID: 35526271 PMCID: PMC9197406 DOI: 10.1111/acel.13626] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/13/2022] [Accepted: 04/21/2022] [Indexed: 01/20/2023] Open
Abstract
The proportion of humans suffering from age‐related diseases is increasing around the world, and creative solutions are needed to promote healthy longevity. Recent work has clearly shown that a calorie is not just a calorie—and that low protein diets are associated with reduced mortality in humans and promote metabolic health and extended lifespan in rodents. Many of the benefits of protein restriction on metabolism and aging are the result of decreased consumption of the three branched‐chain amino acids (BCAAs), leucine, isoleucine, and valine. Here, we discuss the emerging evidence that BCAAs are critical modulators of healthy metabolism and longevity in rodents and humans, as well as the physiological and molecular mechanisms that may drive the benefits of BCAA restriction. Our results illustrate that protein quality—the specific composition of dietary protein—may be a previously unappreciated driver of metabolic dysfunction and that reducing dietary BCAAs may be a promising new approach to delay and prevent diseases of aging.
Collapse
Affiliation(s)
- Michaela E. Trautman
- Department of Medicine University of Wisconsin‐Madison Madison Wisconsin USA
- William S. Middleton Memorial Veterans Hospital Madison Wisconsin USA
- Interdepartmental Graduate Program in Nutritional Sciences University of Wisconsin‐Madison Madison Wisconsin USA
| | - Nicole E. Richardson
- Department of Medicine University of Wisconsin‐Madison Madison Wisconsin USA
- William S. Middleton Memorial Veterans Hospital Madison Wisconsin USA
- Endocrinology and Reproductive Physiology Graduate Training Program University of Wisconsin‐Madison Madison Wisconsin USA
| | - Dudley W. Lamming
- Department of Medicine University of Wisconsin‐Madison Madison Wisconsin USA
- William S. Middleton Memorial Veterans Hospital Madison Wisconsin USA
- Endocrinology and Reproductive Physiology Graduate Training Program University of Wisconsin‐Madison Madison Wisconsin USA
| |
Collapse
|
28
|
Abstract
The cardiomyocyte circadian clock temporally governs fundamental cellular processes, leading to 24-h rhythms in cardiac properties (such as electrophysiology and contractility). The importance of this cell-autonomous clock is underscored by reports that the disruption of the mechanism leads to adverse cardiac remodeling and heart failure. In healthy non-stressed mice, the cardiomyocyte circadian clock modestly augments both cardiac protein synthesis (~14%) and mass (~11%) at the awake-to-sleep transition (relative to their lowest values in the middle of the awake period). However, the increased capacity for cardiac growth at the awake-to-sleep transition exacerbates the responsiveness of the heart to pro-hypertrophic stimuli/stresses (e.g., adrenergic stimulation, nutrients) at this time. The cardiomyocyte circadian clock orchestrates time-of-day-dependent rhythms in cardiac growth through numerous mechanisms. Both ribosomal RNA (e.g., 28S) and the PI3K/AKT/mTOR/S6 signaling axis are circadian regulated, peaking at the awake-to-sleep transition in the heart. Conversely, the negative regulators of translation (including PER2, AMPK, and the integrated stress response) are elevated in the middle of the awake period in a coordinated fashion. We speculate that persistent circadian governance of cardiac growth during non-dipping/nocturnal hypertension, sleep apnea, and/or shift work may exacerbate left ventricular hypertrophy and cardiac disease development, highlighting a need for the advancement of chronotherapeutic interventions.
Collapse
Affiliation(s)
| | - Martin E. Young
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| |
Collapse
|
29
|
Dolci S, Mannino L, Bottani E, Campanelli A, Di Chio M, Zorzin S, D'Arrigo G, Amenta A, Segala A, Paglia G, Denti V, Fumagalli G, Nisoli E, Valerio A, Verderio C, Martano G, Bifari F, Decimo I. Therapeutic Induction of Energy Metabolism Reduces Neural Tissue Damage and Increases Microglia Activation in Severe Spinal Cord Injury. Pharmacol Res 2022; 178:106149. [PMID: 35240272 DOI: 10.1016/j.phrs.2022.106149] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/16/2022] [Accepted: 02/26/2022] [Indexed: 01/11/2023]
Abstract
Neural tissue has high metabolic requirements. Following spinal cord injury (SCI), the damaged, tissue suffers from a severe metabolic impairment, which aggravates axonal degeneration and, neuronal loss. Impaired cellular energetic, tricarboxylic acid (TCA) cycle and oxidative, phosphorylation metabolism in neuronal cells has been demonstrated to be a major cause of neural tissue death and regeneration failure following SCI. Therefore, rewiring the spinal cord cell metabolism may be an innovative therapeutic strategy for the treatment of SCI. In this study, we evaluated the therapeutic effect of the recovery of oxidative metabolism in a mouse model of severe contusive SCI. Oral administration of TCA cycle intermediates, co-factors, essential amino acids, and branched-chain amino acids was started 3 days post-injury and continued until the end of the experimental procedures. Metabolomic, immunohistological, and biochemical analyses were performed on the injured spinal cord sections. Administration of metabolic precursors enhanced spinal cord oxidative metabolism. In line with this metabolic shift, we observed the activation of the mTORC1 anabolic pathway, the increase in mitochondrial mass, and ROS defense which effectively prevented the injury-induced neural cell apoptosis in treated animals. Consistently, we found more choline acetyltransferase (ChAT)-expressing motor neurons and increased neurofilament positive corticospinal axons in the spinal cord parenchyma of the treated mice. Interestingly, oral administration of the metabolic precursors increased the number of activated microglia expressing the CD206 marker suggestive of a, pro-resolutive, M2-like phenotype. These molecular and histological modifications observed in treated animals ultimately led to a significant, although partial, improvement of the motor functions. Our data demonstrate that rewiring the cellular metabolism can represent an effective strategy to treat SCI.
Collapse
Affiliation(s)
- Sissi Dolci
- Department of Diagnostic and Public Health, Section of Pharmacology, University of Verona, 37134, Italy
| | - Loris Mannino
- Department of Diagnostic and Public Health, Section of Pharmacology, University of Verona, 37134, Italy
| | - Emanuela Bottani
- Department of Diagnostic and Public Health, Section of Pharmacology, University of Verona, 37134, Italy
| | - Alessandra Campanelli
- Department of Diagnostic and Public Health, Section of Pharmacology, University of Verona, 37134, Italy
| | - Marzia Di Chio
- Department of Diagnostic and Public Health, Section of Pharmacology, University of Verona, 37134, Italy
| | - Stefania Zorzin
- Department of Diagnostic and Public Health, Section of Pharmacology, University of Verona, 37134, Italy
| | | | - Alessia Amenta
- Laboratory of Cell Metabolism and Regenerative Medicine, Department of Medical Biotechnology and Translational Medicine, University of Milan, 20133, Italy
| | - Agnese Segala
- Department of Molecular and Translational Medicine, University of Brescia, 25121, Italy
| | - Giuseppe Paglia
- School of Medicine and Surgery, Università degli Studi di Milano-Bicocca, 20126, Italy
| | - Vanna Denti
- School of Medicine and Surgery, Università degli Studi di Milano-Bicocca, 20126, Italy
| | - Guido Fumagalli
- Department of Diagnostic and Public Health, Section of Pharmacology, University of Verona, 37134, Italy
| | - Enzo Nisoli
- Center for Study and Research on Obesity, Department of Medical Biotechnology and Translational Medicine, University of Milan, 20133, Italy
| | - Alessandra Valerio
- Department of Molecular and Translational Medicine, University of Brescia, 25121, Italy
| | | | | | - Francesco Bifari
- Laboratory of Cell Metabolism and Regenerative Medicine, Department of Medical Biotechnology and Translational Medicine, University of Milan, 20133, Italy.
| | - Ilaria Decimo
- Department of Diagnostic and Public Health, Section of Pharmacology, University of Verona, 37134, Italy.
| |
Collapse
|
30
|
Shi X, Qiu H. New Insights Into Energy Substrate Utilization and Metabolic Remodeling in Cardiac Physiological Adaption. Front Physiol 2022; 13:831829. [PMID: 35283773 PMCID: PMC8914108 DOI: 10.3389/fphys.2022.831829] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/10/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiac function highly relies on sufficient energy supply. Perturbations in myocardial energy metabolism play a causative role in cardiac pathogenesis. Accumulating evidence has suggested that modifications of cardiac metabolism are also an essential part of the adaptive responses to various physiological conditions in the heart to meet specific energy needs. The review highlighted some new studies on basic myocardial energy substrate metabolism and updated recent findings regarding cardiac metabolic remodeling and their associated mechanisms under physiological conditions, including exercise and cardiac development. Studying basic metabolic profiles in the heart in these conditions can contribute to understanding the significance of metabolic regulation in the heart during physiological adaption and gaining further insights into the maladaptive metabolic changes associated with cardiac pathogenesis, thus opening up new avenues to exploring novel therapeutic strategies in cardiac diseases.
Collapse
|
31
|
Sonkar R, Berry R, Latimer MN, Prabhu SD, Young ME, Frank SJ. Augmented Cardiac Growth Hormone Signaling Contributes to Cardiomyopathy Following Genetic Disruption of the Cardiomyocyte Circadian Clock. Front Pharmacol 2022; 13:836725. [PMID: 35250583 PMCID: PMC8888912 DOI: 10.3389/fphar.2022.836725] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/21/2022] [Indexed: 11/25/2022] Open
Abstract
Circadian clocks regulate numerous biological processes, at whole body, organ, and cellular levels. This includes both hormone secretion and target tissue sensitivity. Although growth hormone (GH) secretion is time-of-day-dependent (increased pulse amplitude during the sleep period), little is known regarding whether circadian clocks modulate GH sensitivity in target tissues. GH acts in part through induction of insulin-like growth factor 1 (IGF1), and excess GH/IGF1 signaling has been linked to pathologies such as insulin resistance, acromegaly, and cardiomyopathy. Interestingly, genetic disruption of the cardiomyocyte circadian clock leads to cardiac adverse remodeling, contractile dysfunction, and reduced lifespan. These observations led to the hypothesis that the cardiomyopathy observed following cardiomyocyte circadian clock disruption may be secondary to chronic activation of cardiac GH/IGF1 signaling. Here, we report that cardiomyocyte-specific BMAL1 knockout (CBK) mice exhibit increased cardiac GH sensitivity, as evidenced by augmented GH-induced STAT5 phosphorylation (relative to littermate controls) in the heart (but not in the liver). Moreover, Igf1 mRNA levels are approximately 2-fold higher in CBK hearts (but not in livers), associated with markers of GH/IGF1 signaling activation (e.g., p-ERK, p-mTOR, and p-4EBP1) and adverse remodeling (e.g., cardiomyocyte hypertrophy and interstitial fibrosis). Genetic deletion of one allele of the GH receptor (GHR) normalized cardiac Igf1 levels in CBK hearts, associated with a partial normalization of adverse remodeling. This included attenuated progression of cardiomyopathy in CBK mice. Collectively, these observations suggest that excessive cardiac GH/IGF1 signaling contributes toward cardiomyopathy following genetic disruption of the cardiomyocyte circadian clock.
Collapse
Affiliation(s)
- Ravi Sonkar
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ryan Berry
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mary N. Latimer
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Sumanth D. Prabhu
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Cardiology Section, Birmingham VAMC Medical Service, Birmingham, AL, United States
- Division of Cardiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Martin E. Young
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Stuart J. Frank
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Endocrinology Section, Birmingham VAMC Medical Service, Birmingham, AL, United States
| |
Collapse
|
32
|
Gao C, Yu J, Wang Y. Branched chain amino acids in cardiac growth and pathology - timing is everything. J Mol Cell Cardiol 2021; 159:14-15. [PMID: 34119505 PMCID: PMC9107373 DOI: 10.1016/j.yjmcc.2021.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 11/20/2022]
Affiliation(s)
- Chen Gao
- Molecular Biology Institute, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, United States of America
| | - Jiayu Yu
- Molecular Biology Institute, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, United States of America
| | - Yibin Wang
- Molecular Biology Institute, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, United States of America.
| |
Collapse
|